Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 43(1): 27-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25318994

ABSTRACT

Cynomolgus monkeys are widely used in preclinical studies during drug development because of their evolutionary closeness to humans, including their cytochrome P450s (P450s). Most cynomolgus monkey P450s are almost identical (≥90%) to human P450s; however, CYP2C76 has low sequence identity (approximately 80%) to any human CYP2Cs. Although CYP2C76 has no ortholog in humans and is partly responsible for species differences in drug metabolism between cynomolgus monkeys and humans, a broad evaluation of potential substrates for CYP2C76 has not yet been conducted. In this study, a screening of 89 marketed compounds, including human CYP2C and non-CYP2C substrates or inhibitors, was conducted to find potential CYP2C76 substrates. Among the compounds screened, 19 chemicals were identified as substrates for CYP2C76, including substrates for human CYP1A2 (7-ethoxyresorufin), CYP2B6 (bupropion), CYP2D6 (dextromethorphan), and CYP3A4/5 (dextromethorphan and nifedipine), and inhibitors for CYP2B6 (sertraline, clopidogrel, and ticlopidine), CYP2C8 (quercetin), CYP2C19 (ticlopidine and nootkatone), and CYP3A4/5 (troleandomycin). CYP2C76 metabolized a wide variety of the compounds with diverse structures. Among them, bupropion and nifedipine showed high selectivity to CYP2C76. As for nifedipine, CYP2C76 formed methylhydroxylated nifedipine, which was not produced by monkey CYP2C9, CYP2C19, or CYP3A4, as identified by mass spectrometry and estimated by a molecular docking simulation. This unique oxidative metabolite formation of nifedipine could be one of the selective marker reactions of CYP2C76 among the major CYP2Cs and CYP3As tested. These results suggest that monkey CYP2C76 contributes to bupropion hydroxylation and formation of different nifedipine oxidative metabolites as a result of its relatively large substrate cavity.


Subject(s)
Bupropion/metabolism , Cytochrome P-450 Enzyme System/metabolism , Macaca fascicularis/metabolism , Nifedipine/metabolism , Oxidoreductases/metabolism , Animals , Humans , Hydroxylation/physiology , Molecular Docking Simulation/methods
2.
Xenobiotica ; 43(12): 1037-42, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23679834

ABSTRACT

Caffeine (1,3,7-trimethylxanthine) is a phenotyping substrate for human cytochrome P450 1A2. 3-N-Demethylation of caffeine is the main human metabolic pathway, whereas monkeys extensively mediate the 7-N-demethylation of caffeine to form pharmacological active theophylline. Roles of monkey P450 enzymes in theophylline formation from caffeine were investigated using individual monkey liver microsomes and 14 recombinantly expressed monkey P450 enzymes, and the results were compared with those for human P450 enzymes. Caffeine 7-N-demethylation activity in microsomes from 20 monkey livers was not strongly inhibited by α-naphthoflavone, quinidine or ketoconazole, and was roughly correlated with diclofenac 4'-hydroxylation activities. Monkey P450 2C9 had the highest activity for caffeine 7-N-demethylation. Kinetic analysis revealed that monkey P450 2C9 had a high Vmax/Km value for caffeine 7-N-demethylation, comparable to low Km value for monkey liver microsomes. Caffeine could dock favorably with monkey P450 2C9 modeled for 7-N-demethylation and with human P450 1A2 for 3-N-demethylation. The primary metabolite theophylline was oxidized to 8-hydroxytheophylline in similar ways by liver microsomes and by recombinant P450s in both humans and monkeys. These results collectively suggest a high activity for monkey liver P450 2C9 toward caffeine 7-N-demethylation, whereas, in humans, P450 1A2-mediated caffeine 3-N-demethylation is dominant.


Subject(s)
Caffeine/metabolism , Cytochrome P-450 Enzyme System/metabolism , Haplorhini/metabolism , Liver/enzymology , Theophylline/metabolism , Animals , Caffeine/chemistry , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Humans , Hydroxylation/drug effects , Liver/drug effects , Metabolic Networks and Pathways/drug effects , Methylation/drug effects , Microsomes, Liver/enzymology , Molecular Docking Simulation , Oxidation-Reduction/drug effects , Recombinant Proteins/metabolism , Theophylline/chemistry
3.
Biochem Pharmacol ; 84(12): 1691-5, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23041648

ABSTRACT

Cynomolgus monkeys are widely used as primate models in preclinical studies. However, some differences are occasionally seen between monkeys and humans in the activities of cytochrome P450 enzymes. R- and S-warfarin are model substrates for stereoselective oxidation in humans. In this current research, the activities of monkey liver microsomes and 14 recombinantly expressed monkey cytochrome P450 enzymes were analyzed with respect to R- and S-warfarin 6- and 7-hydroxylation. Monkey liver microsomes efficiently mediated both R- and S-warfarin 7-hydroxylation, in contrast to human liver microsomes, which preferentially catalyzed S-warfarin 7-hydroxylation. R-Warfarin 7-hydroxylation activities in monkey liver microsomes were not inhibited by α-naphthoflavone or ketoconazole, and were roughly correlated with P450 2C19 levels and flurbiprofen 4-hydroxylation activities in microsomes from 20 monkey livers. In contrast, S-warfarin 7-hydroxylation activities were not correlated with the four marker drug oxidation activities used. Among the 14 recombinantly expressed monkey P450 enzymes tested, P450 2C19 had the highest activities for R- and S-warfarin 7-hydroxylations. Monkey P450 3A4 and 3A5 slowly mediated R- and S-warfarin 6-hydroxylations. Kinetic analysis revealed that monkey P450 2C19 had high V(max) and low K(m) values for R-warfarin 7-hydroxylation, comparable to those for monkey liver microsomes. Monkey P450 2C19 also mediated S-warfarin 7-hydroxylation with V(max) and V(max)/K(m) values comparable to those for recombinant human P450 2C9. R-warfarin could dock favorably into monkey P450 2C19 modeled. These results collectively suggest high activities for monkey liver P450 2C19 toward R- and S-warfarin 6- and 7-hydroxylation in contrast to the saturation kinetics of human P450 2C9-mediated S-warfarin 7-hydroxylation.


Subject(s)
Anticoagulants/metabolism , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Warfarin/metabolism , Animals , Haplorhini , Humans , Hydroxylation , Molecular Docking Simulation , Stereoisomerism
4.
Biol Pharm Bull ; 35(4): 473-80, 2012.
Article in English | MEDLINE | ID: mdl-22466549

ABSTRACT

Infection-associated inflammation can alter the expression levels and functions of cytochrome P450s (CYPs). Cyp gene expression is regulated by the activation of several nuclear receptors, including pregnane X receptor (PXR), constitutive androstane receptor (CAR), and aryl hydrocarbon receptor (AhR). These receptors can be activated by xenobiotics, including medicines. Here, to study the xenobiotic-induced fluctuations in CYP during inflammation, we examined the effect of lipopolysaccharide (LPS) treatment on the level of mRNAs encoding hepatic CYPs induced by xenobiotic-activated nuclear receptors, in mice. Both the mRNA induction of Cyp genes and the metabolic activities of CYP proteins were examined. LPS treatment caused a significant decrease in the induced expression of the mRNAs for Cyp3a11, 2c29, 2c55, and 1a2, but not for Cyp2b10. To assess the CYP enzymatic activities, CYP3A-mediated testosterone 6ß-hydroxylation and the intrinsic clearance (CL(int)) of nifedipine in liver microsomes were measured in mice treated with the xenobiotic pregnenolone-16alpha-carbonitrile (PCN) with or without LPS administration. Both assays revealed that the CYP3A activity, which was induced by PCN, declined significantly after LPS treatment, and this decline correlated with the Cyp3a11 mRNA level. In addition, we found that the mRNAs for interleukin (IL)-1ß and tumor necrosis factor (TNF) α were increased after treatment with LPS plus xenobiotics. Our findings demonstrated that LPS treatment reduces the PXR- and AhR-mediated, and possibly CAR-mediated Cyp gene expression and further suggest that these decreases are dependent on inflammatory cytokines in the liver.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic/drug effects , Lipopolysaccharides/pharmacology , Xenobiotics/pharmacology , Animals , Benzo(a)pyrene/pharmacology , Constitutive Androstane Receptor , Inflammation/chemically induced , Inflammation/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Pregnane X Receptor , Pregnenolone Carbonitrile/pharmacology , Pyridines/pharmacology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/agonists , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Steroid/agonists
5.
Curr Drug Metab ; 11(2): 142-52, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20359284

ABSTRACT

Cynomolgus monkey is used in the study of drug metabolism and toxicity due to its evolutionary closeness to human as compared with other non-human primate species. However, it has become certain that drug metabolism in monkeys is different than in humans. Such species differences have not been fully investigated at a molecular level largely due to the scarcity of information on drug-metabolizing enzyme genes. In cynomolgus monkey, we have identified cDNAs for 21 kinds of cytochromes P450 (CYPs), among which CYP2C76 does not correspond to any human CYP isozymes and is partly responsible for the difference in pitavastatin metabolism between cynomolgus monkey and human. In cynomolgus monkey CYP2C76, we identified numerous genetic variants including a null genotype. Heterozygotes for this null genotype are expected to be poor metabolizers in CYP2C76-mediated drug metabolism. To provide new clues to CYP2C76 function, here, we have taken advantage of sequence information that has been recently deposited to public databases to assess the presence of CYP2C76 orthologs in primate species. In this assessment, we found the CYP2C76 cDNA sequence in rhesus monkey, and a gene sequence highly homologous to cynomolgus monkey CYP2C76 in the marmoset and orangutan genomes, raising the possibility that CYP2C76 could also play a role in these primate species. This review paper gives an overview of CYP2C76 from isolation to molecular characterization, and its implication in drug metabolism.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Macaca mulatta/genetics , Macaca mulatta/metabolism , Multigene Family/genetics , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Amino Acid Sequence , Animals , Callithrix/genetics , Callithrix/metabolism , Cytochrome P-450 Enzyme System/isolation & purification , Cytochrome P-450 Enzyme System/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Macaca fascicularis/genetics , Macaca fascicularis/metabolism , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Molecular Sequence Data , Pongo/genetics , Pongo/metabolism
6.
Mol Pharmacol ; 70(2): 477-86, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16648389

ABSTRACT

Monkeys are widely used as a primate model to study drug metabolism because they generally show a metabolic pattern similar to humans. However, the paucity of information on cytochrome P450 (P450) genes has hampered a deep understanding of drug metabolism in the monkey. In this study, we report identification of the CYP2C76 cDNA newly identified in cynomolgus monkey and characterization of this CYP2C along with cynomolgus CYP2C20, CYP2C43, and CYP2C75. The CYP2C76 cDNA contains the open reading frame encoding a protein of 489 amino acids that are only approximately 80% identical to any human or monkey P450 cDNAs. Gene and protein expression of CYP2C76 was confirmed in the liver of cynomolgus and rhesus monkeys but not in humans or the great apes. Moreover, CYP2C76 is located at the end of the CYP2C gene cluster in the monkey genome, the region of which corresponds to the intergenic region adjacent to the CYP2C cluster in the human genome, strongly indicating that this gene does not have the ortholog in humans. Among the four CYP2C genes expressing predominantly in the liver, the expression level of CYP2C76 was the greatest, suggesting that CYP2C76 is a major CYP2C in the monkey liver. Assays for the capacity of CYP2C76 to metabolize drugs using several substrates typical for human CYP2Cs revealed that CYP2C76 showed unique metabolic activity. These results suggest that CYP2C76 contributes to overall drug-metabolizing activity in the monkey liver and might account for species difference occasionally seen in drug metabolism between monkeys and humans.


Subject(s)
Cytochrome P-450 Enzyme System/physiology , Liver/enzymology , Macaca fascicularis/metabolism , Testosterone/metabolism , Tolbutamide/metabolism , Amino Acid Sequence , Animals , Chromosome Mapping , Cytochrome P-450 Enzyme System/genetics , DNA, Complementary/isolation & purification , Female , Humans , Immunoblotting , Immunohistochemistry , Male , Molecular Sequence Data
7.
Pharm Res ; 23(3): 506-12, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16388406

ABSTRACT

PURPOSE: With the growing clinical usage of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins), the number of reports concerning serious drug-drug interaction has been increasing. Because recent studies have shown that conversion between acid and lactone forms occurs in the body, drug-drug interaction should be considered on both acid and lactone forms. Thus, we investigated the inhibitory effects of acid and lactone forms of eight statins, including one recently withdrawn, cerivastatin, and two recently developed, pitavastatin and rosuvastatin, on cytochrome P450 (CYP) 2C8, CYP2C9, and CYP3A4/5 metabolic activities and multidrug resistance protein 1 (MDR1) transporting activity. METHODS: The inhibitory effects of statins on CYP metabolic activities and MDR1 transporting activity were investigated using human liver microsomes and MDR1-overexpressing LLC-GA5-COL150 cells, respectively. RESULTS: The acid forms had minimal inhibitory effects on all CYP activities tested, except for fluvastatin on CYP2C9-mediated tolbutamide 4-hydroxylation (IC50 = 1.7 microM) and simvastatin on CYP3A4/5-mediated paclitaxel 3-hydroxylation (12.0 microM). Lactone forms showed no or minimal inhibitory effects on CYP2C8, CYP2C9, and CYP2C19 activities, except for rosuvastatin on the CYP2C9 activity (20.5 microM), whereas they showed stronger inhibitory effects on the CYP3A4/5 activity with the rank order of atorvastatin (5.6 microM), cerivastatin (8.1 microM), fluvastatin (14.9 microM), simvastatin (15.2 microM), rosuvastatin (20.7 microM), and lovastatin (24.1 microM). Pitavastatin and pravastatin had little inhibitory effect, and a similar order was found also for testosterone 6beta-hydroxylation. MDR1-mediated transport of [3H]digoxin was inhibited only by lactone forms, and the rank order correlated with that of inhibitory effects on both CYP3A4/5 activities. Inhibitory effects on MDR1 activity, and on both CYP3A4/5 activities, could be explained by the lipophilicity; however, a significant correlation was found between the lipophilicity and inhibitory effects on CYP2C8-mediated paclitaxel 6alpha-hydroxylation. CONCLUSIONS: We showed the difference between the acid and lactone forms in terms of drug interaction. The lipophilicity could be one of the important factors for inhibitory effects. In the case of statins, it is important to examine the effects of both forms to understand the events found in clinical settings, including the pleiotropic effects.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cytochrome P-450 Enzyme Inhibitors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Atorvastatin , Cell Line , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Fluorobenzenes/pharmacology , Heptanoic Acids/pharmacology , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Molecular Structure , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rosuvastatin Calcium , Sulfonamides/pharmacology , Swine , Transfection
8.
J Pharm Pharmacol ; 57(10): 1305-11, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16259759

ABSTRACT

The purpose of this study was to gain a better understanding of the transport mechanism of pitavastatin, a novel synthetic HMG-CoA reductase inhibitor. Experiments were performed using oocytes of Xenopus laevis expressing several solute carrier (SLC) transporters and recombinant membrane vesicles expressing several human ABC transporters. The acid form of pitavastatin was shown to be a substrate for human OATP1, OATP2, OATP8, OAT3 and NTCP, and for rat Oatp1 and Oatp4 with relatively low K(m) values. In contrast, these SLC transporters were not involved in the uptake of the lactone form. A significant stimulatory effect was exhibited by pitavastatin lactone, while the acid form did not exhibit ATPase hydrolysis of P-glycoprotein. In the case of breast cancer resistant protein (BCRP), the acid form of pitavastatin is a substrate, whereas the lactone form is not. Taking these results into consideration, several SLC and ABC transporters were identified as critical to the distribution and excretion of pitavastatin in the body. This study showed, for the first time, that acid and lactone forms of pitavastatin differ in substrate activity towards uptake and efflux transporters. These results will potentially contribute to the differences in the pharmacokinetic profiles of pitavastatin.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Organic Anion Transporters/metabolism , Quinolines/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/metabolism , Animals , Carbon Radioisotopes , Dose-Response Relationship, Drug , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Kinetics , Liver-Specific Organic Anion Transporter 1/genetics , Liver-Specific Organic Anion Transporter 1/metabolism , Neoplasm Proteins/metabolism , Oocytes/drug effects , Oocytes/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Quinolines/chemistry , Quinolines/metabolism , Rats , Solute Carrier Organic Anion Transporter Family Member 1B3 , Symporters/genetics , Symporters/metabolism , Xenopus laevis
9.
Arzneimittelforschung ; 54(7): 382-8, 2004.
Article in English | MEDLINE | ID: mdl-15344842

ABSTRACT

To gain a better understanding of the metabolic stability and transport of pitavastatin (CAS 147526-32-7), a new and potent 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, experiments were conducted using human hepatocytes and oocytes of Xenopus laevis expressing human organic anion transporting polypeptide-2 (OATP2), respectively. Almost the entire radioactivity was from the unchanged substance or lactone form in human hepatocytes, and the cytochrome P450 (CYP)-mediated metabolism of pitavastatin was negligible. The results suggested that CYPs are not critically involved in determining the metabolic fate of pitavastatin. The hepatic uptake of pitavastatin reached saturation with a Km of 2.99 +/- 0.79 micromol/L. Also, the uptake of pitavastatin was mediated by OATP2 expressed in oocytes with a Km of 5.53 +/- 1.70 micromol/L. These results indicated that OATP2 plays a major role in the distribution of pitavastatin in liver. Furthermore, to elucidate the increase in the plasma concentration of pitavastatin in a clinical setting, the inhibitory effect of ciclosporin (cyclosporin A, CAS 59865-13-3) on the uptake of pitavastatin was examined. The uptake of pitavastatin was inhibited in the presence of cyclosporin A and the apparent IC50 value was 2.91 +/- 0.78 micromol/L. This result may at least partly explain the drug-drug interaction between pitavastatin and cyclosporin A. In conclusion, the characterization of transporters needs to be taken into account to avoid transporter-mediated drug-drug interaction.


Subject(s)
Hepatocytes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Quinolines/pharmacokinetics , Animals , Biotransformation , Chromatography, High Pressure Liquid , Cryopreservation , Cyclosporine/pharmacology , Estradiol/metabolism , Female , Humans , Indicators and Reagents , Kinetics , Liver-Specific Organic Anion Transporter 1/metabolism , Quinolines/antagonists & inhibitors , Xenopus laevis
10.
Drug Metabol Drug Interact ; 20(1-2): 25-42, 2004.
Article in English | MEDLINE | ID: mdl-15283301

ABSTRACT

A series of studies was conducted to determine the best animal model for human CYP and UGT activities. The investigation focused primarily on the interactions occurring in the CYP- or UGT-mediated metabolism of pitavastatin, and involved in vitro and in vivo experiments. We found that the best animal models for human CYP-mediated hydroxylation and UGT-mediated lactonization of pitavastatin were rats and dogs, respectively. In addition, a large difference in the metabolic properties of pitavastatin was found between monkeys and humans. In the presence of gemfibrozil, the CYP- or UGT-mediated metabolism of pitavastatin was inhibited in vitro. However, gemfibrozil treatment had no inhibitory effect on the AUC of pitavastatin and its lactone form in rats and dogs. We conclude that the plasma level of pitavastatin would not be increased by co-administration of gemfibrozil in humans.


Subject(s)
Enzyme Inhibitors/metabolism , Gemfibrozil/pharmacology , Hypolipidemic Agents/pharmacology , Quinolines/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/immunology , Aryl Hydrocarbon Hydroxylases/metabolism , Dogs , Glucuronosyltransferase/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Immune Sera/immunology , Lactones/metabolism , Macaca fascicularis , Male , Metabolic Clearance Rate , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Animal , Quinolines/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
11.
Nihon Yakurigaku Zasshi ; 123(5): 349-62, 2004 May.
Article in Japanese | MEDLINE | ID: mdl-15118259

ABSTRACT

Today 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) are the most often prescribed drugs among the therapeutics for hypercholesterolemia. Pitavastatin is a novel statin that has been developed entirely in Japan from the biological screening to clinical studies persuing more efficatious statin than hitherto known. Preclinical studies on drug metabolism revealed that pitavastatin is distributed selectively to the liver, excreted into bile without metabolic modification, and efficiently re-circulates to the liver to show a prolonged plasma half-life. In guinea pigs, pitavastatin enhanced hepatic LDL receptor activity and reduced VLDL secretion in a liver perfusion study, and it lowered plasma total cholesterol (TC) levels at 0.3 mg/kg and triglyceride (TG) levels at 1 mg/kg, respectively, and more. From these results, pitavastatin is assumed to lower LDL cholesterol (LDL-C) by promoting LDL receptor expression and further potentiate the cholesterol-lowering effect and exert TG-lowering effect by reducing VLDL secretion. (14)C-Pitavastatin is metabolized with CYP2C9 to 8-hydroxy derivative, but its Vmax /Km was about 2 micro l/min/mg, about 1/8 to 1/100 in comparison to the reported values of other statins, indicating that pitavastatin is hardly metabolized. Also, other human P450 species were not inhibited by pitavastatin. Therefore, pitavastatin is considered to have little interaction with drugs through P450. In the summarized clinical results with 862 patients, pitavastatin lowered TC and LDL-C by 28% and 40%, respectively. There was no difference in the frequency of side effects and no serious adverse effect was observed for pitavastatin. Pitavastatin possesses superior plasma lipid-improving effects, induces little drug interaction, and is expected to make a good contribution to the medication of hypercholesterolemia.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Quinolines/pharmacology , Animals , Arteriosclerosis/drug therapy , Arteriosclerosis/prevention & control , Cholesterol/biosynthesis , Cholesterol, VLDL/metabolism , Citrus paradisi , Clinical Trials as Topic , Cytochrome P-450 Enzyme System/physiology , Drug Interactions , Drug Therapy, Combination , Fenofibrate/therapeutic use , Food-Drug Interactions , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Quinolines/chemistry , Quinolines/pharmacokinetics , Quinolines/therapeutic use , Receptors, LDL/metabolism , Triglycerides/metabolism
12.
Drug Metabol Drug Interact ; 19(3): 161-76, 2003.
Article in English | MEDLINE | ID: mdl-14682608

ABSTRACT

An in vitro study was carried out in order to examine the metabolic basis of the interaction between fibrates and statins. Metabolic inhibition of statins was noted in the presence of gemfibrozil. However, increase in the unchanged form was fairly small for pitavastatin, compared with other statins. Several CYP enzymes were shown to be principally responsible for the metabolism of gemfibrozil in contrast to other fibrates. In the presence of gemfibrozil, a focal point was obtained in Dixon plots, demonstrating that there was inhibition of CYP2C8-, CYP2C9- and CYP3A4-mediated metabolism. We propose that the increase of plasma concentration caused by co-administration of gemfibrozil and statins is at least partially due to CYP-mediated inhibition.


Subject(s)
Gemfibrozil/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Microsomes, Liver/metabolism , Atorvastatin , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/physiology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gemfibrozil/pharmacology , Heptanoic Acids/metabolism , Humans , Pyridines/metabolism , Pyrroles/metabolism , Quinolines/metabolism
13.
Arzneimittelforschung ; 53(10): 701-7, 2003.
Article in English | MEDLINE | ID: mdl-14650362

ABSTRACT

To gain a better understanding of the mechanism of drug-drug interaction between fibrates and statins, several in vitro experiments were performed. On coincubation with several fibrates, pitavastatin (CAS 147526-32-7) did not displace fibrates from their protein binding in human plasma. The presence of gemfibrozil (CAS 25812-30-0) inhibited the metabolism of statins (cerivastatin (CAS 145599-86-6) and atorvastatin (CAS 134523-00-5)) remarkably. However, the increase of the unchanged form was fairly small for pitavastatin. The metabolic profile of gemfibrozil was also investigated. The cytochrome P (CYP) enzyme CYP2C9 plays a major role in the metabolism of gemfibrozil. Gemfibrozil showed a high affinity for CYP enzymes and a relatively high metabolism velocity. Moreover, several inhibitory effects of gemfibrozil on CYP-mediated metabolism were detected--in contrast to other fibrates. Although the mechanism of the drug-drug interaction was not completely clarified, it is suggested that the increase of plasma concentration caused by the co-administration of gemfibrozil and statins is at least partially due to the inhibition of the CYP-mediated metabolism.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Hypolipidemic Agents/pharmacology , Microsomes, Liver/drug effects , Antibodies, Blocking/pharmacology , Blood Proteins/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Humans , In Vitro Techniques , Protein Binding
14.
Arzneimittelforschung ; 53(3): 145-53, 2003.
Article in English | MEDLINE | ID: mdl-12705168

ABSTRACT

To gain a better understanding of drug-drug interaction between various medicinal substances and statins, in vitro experiments using human hepatic microsomes were performed. The metabolic clearance of atorvastatin (CAS 134523-00-5) was about 32 microliters/min/mg protein, some 15-fold greater than that of pitavastatin (CAS 147526-32-7). On co-incubation with several medicinal substances, metabolic inhibition of pitavastatin was negligible in human hepatic microsomes. However, a remarkable metabolic inhibition of atorvastatin was noted in the presence of various medicinal substances. The intrinsic clearance of atorvastatin lactone was 20-fold greater than that of its acid form, whereas no marked difference was noted between pitavastatin and its lactone form. Pitavastatin lactone showed no inhibitory effect on CYP3A4-mediated metabolism of testosterone in contrast to atorvastatin lactone. These results suggest that pitavastatin and its lactone form will be highly unlikely to interact with other drugs in clinical practice.


Subject(s)
Anticholesteremic Agents/adverse effects , Heptanoic Acids/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Pyrroles/adverse effects , Quinolines/adverse effects , Acids/metabolism , Anticholesteremic Agents/pharmacokinetics , Atorvastatin , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Heptanoic Acids/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , In Vitro Techniques , Indicators and Reagents , Lactones/metabolism , Microsomes, Liver/metabolism , Pyrroles/pharmacokinetics , Quinolines/pharmacokinetics
15.
Drug Metab Pharmacokinet ; 18(4): 245-51, 2003.
Article in English | MEDLINE | ID: mdl-15618742

ABSTRACT

To understand the mechanism underlying the highly liver-selective distribution of pitavastatin, uptake experiments were performed using rat hepatocytes. The uptake of pitavastatin into rat hepatocytes is carrier-mediated and involved nonspecific diffusion in the presence of Na(+). The michaelis constant (K(m)) was 26.0 micromol/L, maximal uptake velocity (V(max)) was 3124 pmol/min/mg protein, and non-specific uptake (P(dif)) was 1.16 microL/min/mg protein. There were no remarkable differences in these kinetic parameters between the presence and absence of Na(+). Experiments using metabolic inhibitors revealed that energy-dependent systems contribute to the uptake of pitavastatin in the liver. Some organic anions reduced the uptake into rat hepatocytes in a concentration-dependent manner. The observed rates of inhibition of pitavastatin uptake by BSP, TCA and pravastatin were compared with the predicted rates. The predicted values were calculated, assuming that BSP, TCA and pravastatin inhibit the uptake of pitavastatin in a competitive manner. The observed inhibition by BSP and TCA was similar to that predicted, but the observed inhibition by pravastatin was considerably less than that predicted. In conclusion, most of the pitavastatin taken up into the liver is transported by multiple carrier-mediated transporters such as Na(+)-independent multispecific anion transporters and energy-dependent transporters. In addition, these systems for pitavastatin may have features in common with the BSP and TCA transport system, and may partially involve the pravastatin transport system.

16.
Arzneimittelforschung ; 52(10): 745-53, 2002.
Article in English | MEDLINE | ID: mdl-12442637

ABSTRACT

Pitavastatin (CAS 147526-32-7, NK-104) is a new and very potent competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase and has been approved for treatment of hyperlipoproteinaemia. Pitavastatin has been studied for its effects on hepatic microsomal drug metabolism in rats, and the activities of several drug-metabolizing enzymes have been measured. No induction of the drug metabolizing enzymes (aniline hydroxylase, aminopyrine N-demethylase, 7-ethoxycoumarin O-deethylase and UDP-glucuronic acid transferase) was found in the pitavastatin group compared to the control after the multiple administrations of pitavastatin at the dosage of 1-10 mg/kg per day for 7 days. Based on several different in vitro approaches, it is concluded that CYP2C9 is the enzyme responsible for the metabolism of pitavastatin and no metabolite is present in renal and intestinal microsomes. The CYP2C9 polymorphism was not involved in the pitavastatin metabolism. No inhibitory effect in CYP-mediated metabolism was detected on the tolbutamide 4-hydroxylation (CYP2C9) and testosterone 6 beta-hydroxylation (CYP3A4) in the presence of pitavastatin. The results suggested that pitavastatin did not affect the drug-metabolizing systems.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Quinolines/pharmacokinetics , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Biotransformation , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/biosynthesis , Enzyme Induction/drug effects , Humans , In Vitro Techniques , Liver/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Polymorphism, Genetic/genetics , Rats , Rats, Wistar
17.
Drug Metab Pharmacokinet ; 17(5): 449-56, 2002.
Article in English | MEDLINE | ID: mdl-15618696

ABSTRACT

Pitavastatin is a potent competitive inhibitor of HMG-CoA reductase. In the current study, to elucidate the hepatobiliary excretion of pitavastatin, we investigated the plasma concentration and biliary excretion of (14)C-pitavastatin in EHBR. We also evaluated the distribution of pitavastatin in mdr1a/b knockout mice by whole body autoradiography and quantitative radioassay. In view of the widespread clinical use of pitavastatin and the importance of drug-drug interaction, the inhibitory effect on Pgp-mediated activation of ATPase was also investigated. No marked difference was observed in the plasma concentration and biliary excretion of radioactivity between SDR and EHBR after dosing of (14)C-pitavastatin. Little radioactive transfer into the brain was detected in mdr1a/b knockout mice and the ATPase activity of human Pgp was negligible in the presence of pitavastatin. Moreover, no inhibitory effect on the Pgp-mediated activation of ATPase by verapamil was found in the presence of pitavastatin over a wide concentration range. These results indicated that a cMOAT and Pgp-mediated transport mechanism did not play a major role in the distribution of pitavastatin.

SELECTION OF CITATIONS
SEARCH DETAIL
...